Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 46
1.
Allergy ; 79(2): 445-455, 2024 Feb.
Article En | MEDLINE | ID: mdl-37916710

BACKGROUND: Conventional basophil activation tests (BATs) measure basophil activation by the increased expression of CD63. Previously, fluorophore-labeled avidin, a positively-charged molecule, was found to bind to activated basophils, which tend to expose negatively charged granule constituents during degranulation. This study further compares avidin versus CD63 as basophil activation biomarkers in classifying peanut allergy. METHODS: Seventy subjects with either a peanut allergy (N = 47), a food allergy other than peanut (N = 6), or no food allergy (N = 17) were evaluated. We conducted BATs in response to seven peanut extract (PE) concentrations (0.01-10,000 ng/mL) and four control conditions (no stimulant, anti-IgE, fMLP (N-formylmethionine-leucyl-phenylalanine), and anti-FcεRI). We measured avidin binding and CD63 expression on basophils with flow cytometry. We evaluated logistic regression and XGBoost models for peanut allergy classification and feature identification. RESULTS: Avidin binding was correlated with CD63 expression. Both markers discriminated between subjects with and without a peanut allergy. Although small by percentage, an avidin+ /CD63- cell subset was found in all allergic subjects tested, indicating that the combination of avidin and CD63 could allow a more comprehensive identification of activated basophils. Indeed, we obtained the best classification accuracy (97.8% sensitivity, 96.7% specificity) by combining avidin and CD63 across seven PE doses. Similar accuracy was obtained by combining PE dose of 10,000 ng/mL for avidin and PE doses of 10 and 100 ng/mL for CD63. CONCLUSIONS: Avidin and CD63 are reliable BAT activation markers associated with degranulation. Their combination enhances the identification of activated basophils and improves the classification accuracy of peanut allergy.


Basophil Degranulation Test , Peanut Hypersensitivity , Humans , Peanut Hypersensitivity/diagnosis , Peanut Hypersensitivity/metabolism , Avidin/metabolism , Immunoglobulin E/metabolism , Basophils/metabolism , Flow Cytometry , Arachis , Tetraspanin 30/metabolism
2.
J Allergy Clin Immunol ; 149(1): 262-274, 2022 01.
Article En | MEDLINE | ID: mdl-34051223

BACKGROUND: The etiology of food allergy is poorly understood; mouse models are powerful systems to discover immunologic pathways driving allergic disease. C3H/HeJ mice are a widely used model for the study of peanut allergy because, unlike C57BL/6 or BALB/c mice, they are highly susceptible to oral anaphylaxis. However, the immunologic mechanism of this strain's susceptibility is not known. OBJECTIVE: We aimed to determine the mechanism underlying the unique susceptibility to anaphylaxis in C3H/HeJ mice. We tested the role of deleterious Toll-like receptor 4 (Tlr4) or dedicator of cytokinesis 8 (Dock8) mutations in this strain because both genes have been associated with food allergy. METHODS: We generated C3H/HeJ mice with corrected Dock8 or Tlr4 alleles and sensitized and challenged them with peanut. We then characterized the antibody response to sensitization, anaphylaxis response to both oral and systemic peanut challenge, gut microbiome, and biomarkers of gut permeability. RESULTS: In contrast to C3H/HeJ mice, C57BL/6 mice were resistant to anaphylaxis after oral peanut challenge; however, both strains undergo anaphylaxis with intraperitoneal challenge. Restoring Tlr4 or Dock8 function in C3H/HeJ mice did not protect from anaphylaxis. Instead, we discovered enhanced gut permeability resulting in ingested allergens in the bloodstream in C3H/HeJ mice compared to C57BL/6 mice, which correlated with an increased number of goblet cells in the small intestine. CONCLUSIONS: Our work highlights the potential importance of gut permeability in driving anaphylaxis to ingested food allergens; it also indicates that genetic loci outside of Tlr4 and Dock8 are responsible for the oral anaphylactic susceptibility of C3H/HeJ mice.


Intestinal Mucosa/metabolism , Passive Cutaneous Anaphylaxis , Peanut Hypersensitivity/metabolism , Administration, Oral , Animals , Arachis/immunology , Disease Models, Animal , Female , Gastrointestinal Microbiome , Genetic Predisposition to Disease , Guanine Nucleotide Exchange Factors/genetics , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation , Passive Cutaneous Anaphylaxis/genetics , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/microbiology , Permeability , Species Specificity , Toll-Like Receptor 4/genetics
3.
Sci Rep ; 11(1): 10141, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980880

Evidence has suggested that major peanut allergen Ara h 1 activates dendritic cells (DCs) via interaction with DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin), a C-type lectin receptor, and contributes to development of peanut allergy. Since macrophages, as well as DCs, play a crucial role in innate immunity, we investigated whether natural Ara h 1 (nAra h 1) activates two different subsets of macrophages, human monocyte derived macrophage type 1 (hMDM1: pro-inflammatory model) and type 2 (hMDM2: anti-inflammatory model). hMDM1 and hMDM2 predominantly produced pro-inflammatory cytokines (IL-6 and TNF-α) and an anti-inflammatory cytokine (IL-10) in response to nAra h 1, respectively. hMDM2 took up nAra h 1 and expressed DC-SIGN at higher levels than hMDM1. However, small interfering RNA knockdown of DC-SIGN did not suppress nAra h 1 uptake and nAra h 1-mediated cytokine production in hMDM2. Inhibitors of scavenger receptor class A type I (SR-AI) suppressed the response of hMDM2, but not of hMDM1, suggesting that SR-AI is a major receptor in hMDM2 for nAra h 1 recognition and internalization. nAra h 1 appears to exert stimulatory capacity on DC and macrophages via different receptors. This study advances our understanding how a major peanut allergen interacts with innate immunity.


Allergens/immunology , Antigens, Plant/immunology , Arachis/immunology , Cell Plasticity/immunology , Macrophages/immunology , Membrane Proteins/immunology , Monocytes/immunology , Peanut Hypersensitivity/immunology , Plant Proteins/immunology , Biomarkers , Disease Susceptibility , Humans , Immunophenotyping , Macrophage Activation/immunology , Macrophages/metabolism , Monocytes/metabolism , Peanut Hypersensitivity/diagnosis , Peanut Hypersensitivity/metabolism
4.
Clin Exp Allergy ; 51(5): 666-673, 2021 05.
Article En | MEDLINE | ID: mdl-33721370

BACKGROUND: Children with atopic dermatitis (AD) are often sensitized to food and aeroallergens, but sensitization patterns have not been analysed with biologic measures of disease pathogenicity. OBJECTIVE: We sought to define allergen sensitization grouping(s) using unbiased machine learning and determine their associations with skin filaggrin (FLG) and transepidermal water loss (TEWL) (assesses skin barrier integrity), S100A8 and S100A9 expression (assesses skin inflammation) and AD severity. METHODS: We studied 400 children with AD in the Mechanisms of Progression from Atopic Dermatitis to Asthma in Children (MPAACH) cohort to identify groupings of food and aeroallergen sensitizations. MPAACH is a paediatric AD cohort, aged 1-2, recruited through hospital/community settings between 2016 and 2018. We analysed these groupings' associations with AD biomarkers: skin FLG, S100A8 and S100A9 expression, total IgE, TEWL and AD severity. RESULTS: An unbiased machine learning approach revealed five allergen clusters. The most common cluster (N = 131), SPTPEP, had sensitization to peanut, egg and/or pets. Three low prevalence clusters, which included children with allergen sensitization other than peanut, egg or pets, were combined into SPTOther . SPTNEG included children with no sensitization(s). SPTPEP children had higher median non-lesional TEWL (16.9 g/m2 /h) and IgE (90 kU/L) compared with SPTOTHER (8.8 g/m2 /h and 24 kU/L; p = .01 and p < .001) and SPTNEG (9 g/m2 /h and 26 kU/L; p = .003 and p < .001). SPTPEP children had lower median lesional (0.70) and non-lesional (1.09) FLG expression compared with SPTOTHER (lesional: 0.9; p = .047, non-lesional: 1.78; p = .01) and SPTNEG (lesional: 1.47; p < .001, non-lesional: 2.21; p < .001). There were no differences among groupings in S100A8 or S100A9 expression. CONCLUSIONS AND CLINICAL RELEVANCE: In this largely clinic-based cohort of young children with AD, allergic sensitization to peanut, egg, cat or dog was associated with more severe disease and skin barrier function but not markers of cutaneous inflammation. These data need replicating in a population-based cohort but may have important implications for understanding the interaction between AD and allergic sensitization.


Calgranulin A/metabolism , Calgranulin B/metabolism , Dermatitis, Atopic/immunology , Filaggrin Proteins/metabolism , Skin/metabolism , Water Loss, Insensible , Animals , Cats , Child, Preschool , Dermatitis, Atopic/metabolism , Dogs , Egg Hypersensitivity/immunology , Egg Hypersensitivity/metabolism , Egg Hypersensitivity/physiopathology , Female , Humans , Infant , Machine Learning , Male , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/physiopathology , Pets/immunology , Skin/physiopathology , Skin Tests
5.
Front Immunol ; 11: 594350, 2020.
Article En | MEDLINE | ID: mdl-33584660

Food allergy is a collective term for several immune-mediated responses to food. IgE-mediated food allergy is the best-known subtype. The patients present with a marked diversity of clinical profiles including symptomatic manifestations, threshold reactivity and reaction kinetics. In-vitro predictors of these clinical phenotypes are evasive and considered as knowledge gaps in food allergy diagnosis and risk management. Peanut allergy is a relevant disease model where pioneer discoveries were made in diagnosis, immunotherapy and prevention. This review provides an overview on the immune basis for phenotype variations in peanut-allergic individuals, in the light of future patient stratification along emerging omic-areas. Beyond specific IgE-signatures and basophil reactivity profiles with established correlation to clinical outcome, allergenomics, mass spectrometric resolution of peripheral allergen tracing, might be a fundamental approach to understand disease pathophysiology underlying biomarker discovery. Deep immune phenotyping is thought to reveal differential cell responses but also, gene expression and gene methylation profiles (eg, peanut severity genes) are promising areas for biomarker research. Finally, the study of microbiome-host interactions with a focus on the immune system modulation might hold the key to understand tissue-specific responses and symptoms. The immune mechanism underlying acute food-allergic events remains elusive until today. Deciphering this immunological response shall enable to identify novel biomarker for stratification of patients into reaction endotypes. The availability of powerful multi-omics technologies, together with integrated data analysis, network-based approaches and unbiased machine learning holds out the prospect of providing clinically useful biomarkers or biomarker signatures being predictive for reaction phenotypes.


Allergens/immunology , Arachis/adverse effects , Biomarkers , Immunoglobulin E/immunology , Peanut Hypersensitivity/diagnosis , Peanut Hypersensitivity/etiology , Phenotype , Animals , Genomics/methods , Humans , Microbiota/immunology , Peanut Hypersensitivity/metabolism , Prognosis , Proteomics/methods
7.
J Agric Food Chem ; 66(41): 10855-10863, 2018 Oct 17.
Article En | MEDLINE | ID: mdl-30284821

The 2S albumin Ara h 6 is one of the most important peanut allergens. A post-translationally cleaved Ara h 6 (pAra h 6) was purified from Virginia type peanuts, and the cleavage site was mapped using high-resolution mass spectrometry. Compared to intact Ara h 6, pAra h 6 lacks a 5-amino acid stretch, resembling amino acids 43-47 (UniProt accession number Q647G9) in the nonstructured loop. Consequently, pAra h 6 consists of two chains: an N-terminal chain of approximately 5 kDa and a C-terminal chain of approximately 9 kDa, held together by disulfide bonds. Intermediate post-translationally cleaved products, in which this stretch is cleaved yet still attached to one of the subunits, are also present. The secondary structure and immunoglobulin E (IgE) binding of pAra h 6 resembles that of intact Ara h 6, indicating that the loss of the nonstructured loop is not critical for maintaining the protein structure. Commercially available monoclonal and polyclonal immunoglobulin G (IgG) antibodies directed to Ara h 6 react with both intact Ara h 6 and pAra h 6, suggesting that the involved epitopes are not located in the area that is post-translationally cleaved. No differences between intact Ara h 6 and pAra h 6 in terms of IgE binding were found, suggesting that the area that is post-translationally cleaved is not involved in IgE epitopes either. For all main cultivars Runner, Virginia, Valencia, and Spanish, intact Ara h 6 and pAra h 6 occur in peanut at similar levels, indicating that pAra h 6 is a consistent and important contributor to the allergenic potency of peanut.


2S Albumins, Plant/chemistry , 2S Albumins, Plant/isolation & purification , Antigens, Plant/chemistry , Antigens, Plant/isolation & purification , Arachis/chemistry , 2S Albumins, Plant/immunology , Amino Acid Sequence , Amino Acids/chemistry , Antigens, Plant/immunology , Epitopes/immunology , Glycoproteins/chemistry , Glycoproteins/immunology , Glycoproteins/isolation & purification , Humans , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/prevention & control , Plant Proteins/chemistry , Plant Proteins/immunology , Plant Proteins/isolation & purification , Protein Structure, Secondary
8.
Nutrients ; 10(9)2018 Sep 11.
Article En | MEDLINE | ID: mdl-30208580

The oral mucosa is the first immune tissue that encounters allergens upon ingestion of food. We hypothesized that the bio-accessibility of allergens at this stage may be a key determinant for sensitization. Light roasted peanut flour was suspended at various pH in buffers mimicking saliva. Protein concentrations and allergens profiles were determined in the supernatants. Peanut protein solubility was poor in the pH range between 3 and 6, while at a low pH (1.5) and at moderately high pHs (>8), it increased. In the pH range of saliva, between 6.5 and 8.5, the allergens Ara h2 and Ara h6 were readily released, whereas Ara h1 and Ara h3 were poorly released. Increasing the pH from 6.5 to 8.5 slightly increased the release of Ara h1 and Ara h3, but the recovery remained low (approximately 20%) compared to that of Ara h2 and Ara h6 (approximately 100% and 65%, respectively). This remarkable difference in the extraction kinetics suggests that Ara h2 and Ara h6 are the first allergens an individual is exposed to upon ingestion of peanut-containing food. We conclude that the peanut allergens Ara h2 and Ara h6 are quickly bio-accessible in the mouth, potentially explaining their extraordinary allergenicity.


2S Albumins, Plant/metabolism , Antigens, Plant/metabolism , Arachis/metabolism , Glycoproteins/metabolism , Nuts/metabolism , Peanut Hypersensitivity/metabolism , Saliva/metabolism , 2S Albumins, Plant/immunology , Antigens, Plant/immunology , Arachis/immunology , Buffers , Glycoproteins/immunology , Humans , Hydrogen-Ion Concentration , Immunity, Mucosal , Kinetics , Mouth Mucosa/immunology , Mouth Mucosa/metabolism , Nuts/immunology , Peanut Hypersensitivity/immunology , Solubility
9.
Nutrients ; 10(6)2018 Jun 08.
Article En | MEDLINE | ID: mdl-29890625

BACKGROUND: Evidence suggests a relevant role for liver and mitochondrial dysfunction in allergic disease. However, the role of hepatic mitochondrial function in food allergy is largely unknown. We aimed to investigate hepatic mitochondrial dysfunction in a murine model of peanut allergy. METHODS: Three-week-old C3H/HeOuJ mice were sensitized by the oral route with peanut-extract (PNT). We investigated: 1. the occurrence of effective sensitization to PNT by analysing acute allergic skin response, anaphylactic symptoms score, body temperature, serum mucosal mast cell protease-1 (mMCP-1) and anti-PNT immunoglobulin E (IgE) levels; 2. hepatic involvement by analysing interleukin (IL)-4, IL-5, IL-13, IL-10 and IFN-γ mRNA expression; 3. hepatic mitochondrial oxidation rates and efficiency by polarography, and hydrogen peroxide (H2O2) yield, aconitase and superoxide dysmutase activities by spectrophotometry. RESULTS: Sensitization to PNT was demonstrated by acute allergic skin response, anaphylactic symptoms score, body temperature decrease, serum mMCP-1 and anti-peanut IgE levels. Liver involvement was demonstrated by a significant increase of hepatic Th2 cytokines (IL-4, IL-5 and IL-13) mRNA expression. Mitochondrial dysfunction was demonstrated by lower state 3 respiration rate in the presence of succinate, decreased fatty acid oxidation in the presence of palmitoyl-carnitine, increased yield of ROS proven by the inactivation of aconitase enzyme and higher H2O2 mitochondrial release. CONCLUSIONS: We provide evidence of hepatic mitochondrial dysfunction in a murine model of peanut allergy. These data could open the way to the identification of new mitochondrial targets for innovative preventive and therapeutic strategies against food allergy.


Allergens/immunology , Arachis/immunology , Energy Metabolism , Liver/immunology , Mitochondria, Liver/immunology , Peanut Hypersensitivity/immunology , Plant Proteins/immunology , Th2 Cells/immunology , Animals , Disease Models, Animal , Female , Immunoglobulin E/blood , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukin-5/genetics , Interleukin-5/immunology , Interleukin-5/metabolism , Liver/metabolism , Mice, Inbred C3H , Mitochondria, Liver/metabolism , Oxidation-Reduction , Oxidative Stress , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Th2 Cells/metabolism , Up-Regulation
10.
Int Arch Allergy Immunol ; 177(1): 35-39, 2018.
Article En | MEDLINE | ID: mdl-29879702

Roasting has been implicated in the increase of peanut allergenicity due to the chemical reactions that occur during the process. However, this increase is not fully understood, and little information is available regarding the role of roasted peanut allergens in the initial phase of allergy, where dendritic cells (DCs) play a key role. We sought to analyze differences in the internalization of Ara h 3 from raw and roasted peanut by immature monocyte-derived DCs (MDDCs) and the implication of the mannose receptor in the uptake. Ara h 3 was purified from raw and roasted peanut (Ara h 3-raw and Ara h 3-roas) and labeled with a fluorescent dye. The labeled allergens were added to MDDCs obtained from 7 donors and internalization was analyzed after 10, 30, and 120 min by flow cytometry. In parallel, mannan, which blocks the mannose receptor, was added 30 min before adding the labeled allergens. Results showed that the internalization of Ara h 3-roas by MDDCs was significantly increased at every time point. However, the increase in the internalization of Ara h 3-raw was only significant after 2 h of incubation. Ara h 3-roas had an enhanced capacity to be internalized by MDDCs in comparison with Ara h 3-raw at every time point. Blocking the mannose receptor decreased the internalization of Ara h 3-roas but not Ara h 3-raw. In conclusion, the internalization of Ara h 3-roas by the MDDCs is enhanced when compared to Ara h 3-raw, and the mannose receptor might be implicated in this enhancement.


Antigens, Plant/immunology , Arachis/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Plant Proteins/immunology , Cell Differentiation , Cells, Cultured , Dendritic Cells/cytology , Endocytosis , Humans , Immunoglobulin E/immunology , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/metabolism , Receptors, Cell Surface/metabolism
11.
Allergy ; 73(1): 137-144, 2018 Jan.
Article En | MEDLINE | ID: mdl-28686296

BACKGROUND: Allergy can be diagnosed using basophil tests. Several methods measuring basophil activation are available. This study aimed at comparing basophil activation test (BAT), histamine release assay (HR), and passive sensitization histamine release assay (passive HR) in the diagnosis of peanut allergy. METHODS: BAT, HR, and passive HR were performed on 11 peanut-allergic and 14 nonallergic subjects. Blood was incubated with peanut extract or anti-IgE and tests were performed as follows: BAT-CD63 upregulation was assessed by flow cytometry; HR-released histamine was quantified by a glass fiber-based fluorometric method; passive HR-IgE-stripped donor basophils were incubated with participants' serum and histamine release was quantified as HR. RESULTS: CDsens, a measure of basophil allergen sensitivity, was significantly higher for BAT (80.1±17.4) compared to HR (23.4±10.31) and passive HR (11.1±2.0). BAT, HR, and passive HR had a clinical sensitivity of 100%, 100%, and 82% and specificity of 100%, 100%, and 100%, respectively, when excluding inconclusive results. BAT identified 11 of 11 allergic patients, HR 10, and passive HR 9. Likewise, BAT recognized 12 of 14 nonallergic subjects, HR 10, and passive HR 13. However, the tests' diagnostic performances were not statistically different. Interestingly, nonreleasers in HR but not in BAT had lower basophil count compared to releasers (249 vs 630 counts/min). CONCLUSION: BAT displayed a significantly higher CDsens compared to HR and passive HR. The basophil tests' diagnostic performances were not significantly different. Still, BAT could diagnose subjects with low basophil number in contrast to HR.


Basophils/immunology , Basophils/metabolism , Histamine Release , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/metabolism , Adolescent , Adult , Allergens/immunology , Antigens, CD/metabolism , Antigens, Plant/immunology , Biomarkers , Case-Control Studies , Female , Humans , Immunization , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunophenotyping , Male , Middle Aged , Peanut Hypersensitivity/diagnosis , Reproducibility of Results , Skin Tests , Young Adult
12.
Allergol Int ; 67(1): 24-31, 2018 Jan.
Article En | MEDLINE | ID: mdl-29233462

Therapeutic strategy in late 20th century to prevent allergic diseases was derived from a conceptual framework of allergens elimination which was as same as that of coping with them after their onset. Manifold trials were implemented; however, most of them failed to verify the effectiveness of their preventive measures. Recent advancement of epidemiological studies and cutaneous biology revealed epidermal barrier dysfunction plays a major role of allergen sensitization and development of atopic dermatitis which ignites the inception of allergy march. For this decade, therapeutic strategy to prevent the development of food allergy has been confronted with a paradigm shift from avoidance and delayed introduction of allergenic foods based on the theoretical concept to early introduction of them based on the clinical and epidemiological evidences. Especially, prevention of peanut allergy and egg allergy has been established with the highest evidence verified by randomized controlled trials, although application in clinical practice should be done with attention. This paradigm shift concerning food allergy was also due to the discovery of cutaneous sensitization risk of food allergens for an infant with eczema revealed by prospective studies. Here we have recognized the increased importance of prevention of eczema/atopic dermatitis in infancy. Two randomized controlled trials using emollients showed successful results in prevention of atopic dermatitis in infancy; however, longer term safety and prognosis including allergy march should be pursued. To establish more fundamental strategy for prevention of the development of allergy, further studies clarifying the mechanisms of interaction between barrier dysfunction and microbial milieu are needed with macroscope to understand the relationship between allergic diseases and a diversity of environmental influences.


Dermatitis, Atopic/immunology , Dermatitis, Atopic/prevention & control , Egg Hypersensitivity/immunology , Egg Hypersensitivity/prevention & control , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/prevention & control , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Dermis/immunology , Dermis/metabolism , Dermis/pathology , Egg Hypersensitivity/metabolism , Egg Hypersensitivity/pathology , Humans , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/pathology , Randomized Controlled Trials as Topic
13.
J Allergy Clin Immunol ; 141(3): 991-1001, 2018 03.
Article En | MEDLINE | ID: mdl-29030101

BACKGROUND: Peanut allergy (PA) is a complex disease with both environmental and genetic risk factors. Previously, PA loci were identified in filaggrin (FLG) and HLA in candidate gene studies, and loci in HLA were identified in a genome-wide association study and meta-analysis. OBJECTIVE: We sought to investigate genetic susceptibility to PA. METHODS: Eight hundred fifty cases and 926 hyper-control subjects and more than 7.8 million genotyped and imputed single nucleotide polymorphisms (SNPs) were analyzed in a genome-wide association study to identify susceptibility variants for PA in the Canadian population. A meta-analysis of 2 phenotypes (PA and food allergy) was conducted by using 7 studies from the Canadian, American (n = 2), Australian, German, and Dutch (n = 2) populations. RESULTS: An SNP near integrin α6 (ITGA6) reached genome-wide significance with PA (P = 1.80 × 10-8), whereas SNPs associated with Src kinase-associated phosphoprotein 1 (SKAP1), matrix metallopeptidase 12 (MMP12)/MMP13, catenin α3 (CTNNA3), rho GTPase-activating protein 24 (ARHGAP24), angiopoietin 4 (ANGPT4), chromosome 11 open reading frame (C11orf30/EMSY), and exocyst complex component 4 (EXOC4) reached a threshold suggestive of association (P ≤ 1.49 × 10-6). In the meta-analysis of PA, loci in or near ITGA6, ANGPT4, MMP12/MMP13, C11orf30, and EXOC4 were significant (P ≤ 1.49 × 10-6). When a phenotype of any food allergy was used for meta-analysis, the C11orf30 locus reached genome-wide significance (P = 7.50 × 10-11), whereas SNPs associated with ITGA6, ANGPT4, MMP12/MMP13, and EXOC4 and additional C11orf30 SNPs were suggestive (P ≤ 1.49 × 10-6). Functional annotation indicated that SKAP1 regulates expression of CBX1, which colocalizes with the EMSY protein coded by C11orf30. CONCLUSION: This study identifies multiple novel loci as risk factors for PA and food allergy and establishes C11orf30 as a risk locus for both PA and food allergy. Multiple genes (C11orf30/EMSY, SKAP1, and CTNNA3) identified by this study are involved in epigenetic regulation of gene expression.


Epigenesis, Genetic , Genetic Loci , Genome-Wide Association Study , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Peanut Hypersensitivity/genetics , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Chromobox Protein Homolog 5 , Female , Filaggrin Proteins , Humans , Male , Peanut Hypersensitivity/epidemiology , Peanut Hypersensitivity/metabolism , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Risk Factors , alpha Catenin/biosynthesis , alpha Catenin/genetics
14.
J Allergy Clin Immunol ; 141(1): 202-213.e8, 2018 01.
Article En | MEDLINE | ID: mdl-28689791

BACKGROUND: Peanut allergy (PA) is a life-threatening condition that lacks regulator-approved treatment. Regulatory T type 1 (TR1) cells are potent suppressors of immune responses and can be induced in vivo upon repeated antigen exposure or in vitro by using tolerogenic dendritic cells. Whether oral immunotherapy (OIT) leads to antigen-specific TR1 cell induction has not been established. OBJECTIVES: We sought to determine whether peanut-specific TR1 cells can be generated in vitro from peripheral blood of patients with PA at baseline or during OIT and whether they are functional compared with peanut-specific TR1 cells induced from healthy control (HC) subjects. METHODS: Tolerogenic dendritic cells were differentiated in the presence of IL-10 from PBMCs of patients with PA and HC subjects pulsed with the main peanut allergens of Arachis hypogaea, Ara h 1 and 2, and used as antigen-presenting cells for autologous CD4+ T cells (CD4+ T cells coincubated with tolerogenic dendritic cells pulsed with the main peanut allergens [pea-T10 cells]). Pea-T10 cells were characterized by the presence of CD49b+ lymphocyte-activation gene 3 (LAG3)+ TR1 cells, antigen-specific proliferative responses, and cytokine production. RESULTS: CD49b+LAG3+ TR1 cells were induced in pea-T10 cells at comparable percentages from HC subjects and patients with PA. Despite their antigen specificity, pea-T10 cells of patients with PA with or without OIT, as compared with those of HC subjects, were not anergic and had high TH2 cytokine production upon peanut-specific restimulation. CONCLUSIONS: Peanut-specific TR1 cells can be induced from HC subjects and patients with PA, but those from patients with PA are functionally defective independent of OIT. The unfavorable TR1/TH2 ratio is discussed as a possible cause of PA TR1 cell impairment.


Allergens/immunology , Antigens, Plant/immunology , Arachis/immunology , Peanut Hypersensitivity/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Biomarkers , Child , Child, Preschool , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Humans , Lymphocyte Activation , Male , Peanut Hypersensitivity/diagnosis , Peanut Hypersensitivity/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Young Adult
15.
Clin Exp Allergy ; 47(10): 1331-1341, 2017 Oct.
Article En | MEDLINE | ID: mdl-28802077

BACKGROUND: Food allergy is an increasingly common health problem in Western populations. Epidemiological studies have suggested both positive and negative associations between food allergy and infection with the gastric bacterium Helicobacter pylori. OBJECTIVE: The objective of this work was to investigate whether experimental infection with H. pylori, or prophylactic treatment with H. pylori-derived immunomodulatory molecules, affects the onset and severity of food allergy, either positively or negatively. METHODS: We infected neonatal C57BL/6 or C3H mice with H. pylori or treated animals with H. pylori components (bacterial lysate or the immunomodulator VacA) and subsequently subjected them to four different protocols for food allergy induction, using either ovalbumin or peanut extract as allergens for sensitization and challenge. Readouts included anaphylaxis scoring, quantification of allergen-specific serum IgE and IgG1 and of the mast cell protease MCPT1, as well as splenic T-helper-2 cell-derived cytokine production. Mesenteric lymph node CD4+ FoxP3+ regulatory T cells were subjected to flow cytometric quantification and sorting followed by qRT-PCR, and to DNA methylation analyses of the Treg-specific demethylated region (TSDR) within the FOXP3 locus. RESULTS: Mice that had been infected with H. pylori or treated with H. pylori-derived immunomodulators showed reduced anaphylaxis upon allergen sensitization and challenge, irrespective of the allergen used. Most of the immunologic assays confirmed a protective effect of H. pylori. CD4+ FoxP3+ T cells were more abundant in protected mice and exhibited a stable Treg phenotype characterized by FOXP3 TSDR demethylation. CONCLUSIONS AND CLINICAL RELEVANCE: Helicobacter pylori confers protection against the anaphylaxis associated with ovalbumin and peanut allergy and affects the epigenome of T cells, thereby promoting stable Treg differentiation and functionality. Prophylactic treatment with H. pylori-derived immunomodulators appears to be a promising strategy for food allergy prevention.


Anaphylaxis/prevention & control , Bacterial Proteins/immunology , Food Hypersensitivity/prevention & control , Helicobacter pylori/immunology , Immunologic Factors/immunology , Allergens/immunology , Anaphylaxis/genetics , Anaphylaxis/immunology , Anaphylaxis/metabolism , Animals , CpG Islands , Cytokines/blood , Cytokines/metabolism , DNA Methylation , Disease Models, Animal , Food Hypersensitivity/genetics , Food Hypersensitivity/immunology , Food Hypersensitivity/metabolism , Immunoglobulin E/immunology , Male , Mice , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/prevention & control , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
16.
Food Chem Toxicol ; 107(Pt A): 88-98, 2017 Sep.
Article En | MEDLINE | ID: mdl-28634114

Stability to proteolytic degradation in the digestive tract is considered a general feature shared by most food allergens. Current digestibility models exclusively utilize purified allergen proteins, neglecting the relevant effects of matrix that occur for foodstuff systems. In the present study, we investigated digestion stability of the major peanut allergens directly in the natural matrix using an in vitro static model that simulates the gastrointestinal digestion including the oral, gastric, duodenal and intestinal (brush border membrane enzymes) phases. Immunogenicity was evaluated by Western Blot using N=8 pooled sera of peanut allergic pediatric subjects. Immunoreactive, large-sized and fragments of Ara h 2, Ara h 6 and Ara h 3 survived hydrolysis as assessed by SDS-PAGE. Smaller resistant peptides mainly arising from Ara h 3 and also Ara h 1 were detected and further identified by LC-high resolution-MS/MS. RP-HPLC purification followed by dot-blot analysis and MS/MS-based identification demonstrated that stable IgE-binding peptides derived from Ara h 3. These results provide a more realistic picture of the potentially allergenic determinants of peanuts that survived the human digestion, taking into account the role of the food matrix, which may significantly affect gastrointestinal breakdown of peanut allergens.


Arachis/chemistry , Immunoglobulin E/immunology , Peanut Hypersensitivity/immunology , Peptides/immunology , Amino Acid Sequence , Antigens, Plant/chemistry , Antigens, Plant/genetics , Antigens, Plant/immunology , Antigens, Plant/metabolism , Arachis/genetics , Arachis/immunology , Arachis/metabolism , Digestion , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Sequence Data , Peanut Hypersensitivity/metabolism , Peptide Mapping , Peptides/chemistry , Peptides/genetics , Tandem Mass Spectrometry
17.
Allergy ; 72(7): 1081-1090, 2017 Jul.
Article En | MEDLINE | ID: mdl-28106273

BACKGROUND: The pathology of allergic diseases involves type 2 immune cells, such as Th2, ILC2, and basophils exerting their effect by production of IL-4, IL-5, and IL-13. However, surface receptors that are specifically expressed on type 2 immune cells are less well documented. The aim of this investigation was to identify surface markers associated with type 2 inflammation. METHODS: Naïve human CD4+ T cells were short-term activated in the presence or absence of IL-4 and analyzed for expression of >300 cell-surface proteins. Ex vivo-isolated peripheral blood mononuclear cells (PBMCs) from peanut-allergic (PA) and nonallergic subjects were stimulated (14-16 h) with peanut extract to detect peanut-specific CD4+ CD154+ T cells. Biopsies were obtained for transcriptomic analysis from healthy controls and patients with extrinsic or intrinsic atopic dermatitis (AD) and psoriasis. RESULTS: Expression analysis of >300 surface proteins enabled identification of IL-4-upregulated surface proteins, such as CD90, CD108, CD109, and CD200R (CD200R1). Additional analysis of in vitro-differentiated Th0, Th1, and Th2 cultures identified CD200R as upregulated on Th2 cells. From ex vivo-isolated PBMCs, we found high expression of CD200R on Th2 and ILC2 cells and basophils. In PA subjects, the peanut-specific Th2 (CD154+ CRTh2+ ) cells expressed more CD200R than the non-allergen-specific Th2 (CD154- CRTh2+ ) cells. Moreover, costaining of CD161 and CD200R identified peanut-specific highly differentiated IL-4+ IL-5+ Th2 cells. Finally, transcriptomic analysis revealed upregulation of CD200R in lesional skin from subjects with an extrinsic AD phenotype compared to healthy skin. CONCLUSION: These results indicate that CD200R expression strongly correlates with Th2 pathology; though, the mechanism is as yet elusive.


Antigens, Surface/genetics , Receptors, Cell Surface/genetics , Th2 Cells/immunology , Th2 Cells/metabolism , Allergens/immunology , Antigens, Surface/metabolism , Basophils/immunology , Basophils/metabolism , Cytokines/metabolism , Gene Expression , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Orexin Receptors , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thy-1 Antigens/metabolism
18.
Clin Exp Allergy ; 47(1): 121-128, 2017 01.
Article En | MEDLINE | ID: mdl-27533916

BACKGROUND: Food allergy has been associated with an increased risk for the development of allergic asthma. Asthma is a risk factor for the development of an anaphylactic response to food allergens. An immunological interplay between sensitization to different allergens in different compartments of the body might be involved. OBJECTIVE: To evaluate the immunological interplay between intragastrical peanut (PE) sensitization and respiratory sensitization to house dust mite (HDM) allergens. METHODS: BALB/c mice were intragastrically sensitized to peanut or sham-sensitized and challenged systemically to PE. Between sensitization and challenge, mice were intranasally exposed to HDM extract or PBS, as a control. The response to HDM (eosinophil recruitment, cytokine response, HDM-specific immunoglobulins and airway hyper-reactivity) and to PE (cytokine response, mast cells in gut, mMCP-1 in serum and body temperature) was assessed. RESULTS: A preceding PE sensitization increased HDM-induced production of IL-4, IL-5, IL-13 and IFNγ in lung-draining lymph nodes and total IgE levels in HDM-sensitized mice. However, recruitment of inflammatory cells to the airways or airway hyper-reactivity was not aggravated in PE/HDM double-sensitized mice. Alternatively, HDM-induced airway inflammation did not significantly affect the immune response or the anaphylactic response to a systemic challenge with peanut. CONCLUSION AND CLINICAL RELEVANCE: Our data show that a preceding peanut sensitization boosted IgE- and HDM-specific Th2 response in the airways in mice. It contributes to the understanding of the underlying immunological mechanism of polysensitization which often occurs in allergic individuals over time.


Allergens/immunology , Antigens, Dermatophagoides/immunology , Arachis/adverse effects , Immunomodulation , Peanut Hypersensitivity/immunology , Pyroglyphidae/immunology , Respiratory Hypersensitivity/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Immunoglobulin E/immunology , Mice , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/pathology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Th2 Cells/immunology , Th2 Cells/metabolism
19.
J Allergy Clin Immunol ; 138(2): 536-543.e4, 2016 08.
Article En | MEDLINE | ID: mdl-27130858

BACKGROUND: Treatments to reverse peanut allergy remain elusive. Current clinical approaches using peanut oral/sublingual immunotherapy are promising, but concerns about safety and long-term benefit remain a barrier to wide use. Improved methods of delivering peanut-specific immunotherapy are needed. OBJECTIVE: We sought to investigate the efficacy and safety of peanut oral immunotherapy using CpG-coated poly(lactic-co-glycolic acid) nanoparticles containing peanut extract (CpG/PN-NPs) in a murine model of peanut allergy. METHODS: C3H/HeJ mice were rendered peanut allergic by means of oral sensitization with peanut and cholera toxin. Mice were then subjected to 4 weekly gavages with CpG/PN-NPs, vehicle (PBS), nanoparticles alone, peanut alone, CpG nanoparticles, or peanut nanoparticles. Untreated mice served as naive controls. After completing therapy, mice underwent 5 monthly oral peanut challenges. Anaphylaxis was evaluated by means of visual assessment of symptom scores and measurement of body temperature and plasma histamine levels. Peanut-specific serum IgE, IgG1, and IgG2a levels were measured by using ELISA, as were cytokine recall responses in splenocyte cultures. RESULTS: Mice with peanut allergy treated with CpG/PN-NPs but not vehicle or other treatment components were significantly protected from anaphylaxis to all 5 oral peanut challenges, as indicated by lower symptom scores, less change in body temperature, and a lower increase of plasma histamine levels. Importantly, CpG/PN-NP treatment did not cause anaphylactic reactions. Treatment was associated with a sustained and significant decrease in peanut-specific IgE/IgG1 levels and an increase in peanut-specific IgG2a levels. Compared with vehicle control animals, peanut recall responses in splenocyte cultures from nanoparticle-treated mice showed significantly decreased levels of TH2 cytokines (IL-4, IL-5, and IL-13) but increased IFN-γ levels in cell supernatants. CONCLUSIONS: Preclinical findings indicate that peanut oral immunotherapy with CpG/PN-NPs might be a valuable strategy for peanut-specific immunotherapy in human subjects.


Allergens/immunology , Arachis/adverse effects , Desensitization, Immunologic , Lactic Acid , Nanoparticles , Peanut Hypersensitivity/immunology , Polyglycolic Acid , Allergens/administration & dosage , Animals , Cytokines/blood , Cytokines/metabolism , Desensitization, Immunologic/methods , Disease Models, Animal , Female , Histamine/blood , Immunization , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Peanut Hypersensitivity/diagnosis , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/therapy , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/immunology , Polylactic Acid-Polyglycolic Acid Copolymer
20.
Allergy ; 71(11): 1561-1574, 2016 11.
Article En | MEDLINE | ID: mdl-27059534

BACKGROUND: Signaling through histamine receptors on dendritic cells (DCs) may be involved in the effector phase of peanut-induced intestinal anaphylaxis. OBJECTIVES: The objective of this study was to determine the role of histamine H1 (H1R) and H4 receptors (H4R) in intestinal allergic responses in a model of peanut allergy. METHODS: Balb/c mice were sensitized and challenged with peanut. During the challenge phase, mice were treated orally with the H1R antagonist, loratadine, and/or the H4R antagonist, JNJ7777120. Bone marrow-derived DCs (BMDCs) were adoptively transferred to nonsensitized WT mice. Symptoms, intestinal inflammation, and mesenteric lymph node and intestine mucosal DCs were assessed. Effects of the drugs on DC chemotaxis, calcium mobilization, and antigen-presenting cell function were measured. RESULTS: Treatment with loratadine or JNJ7777120 individually partially suppressed the development of diarrhea and intestinal inflammation and decreased the numbers of DCs in the mesenteric lymph nodes and lamina propria. Combined treatment with both drugs prevented the development of diarrhea and intestinal inflammation. In vitro, the combination suppressed DC antigen-presenting cell function to T helper cells and DC calcium mobilization and chemotaxis to histamine. CONCLUSION: Blockade of both H1R and H4R in the challenge phase had additive effects in preventing the intestinal consequences of peanut sensitization and challenge. These effects were mediated through the limitation of mesenteric lymph node and intestinal DC accumulation and function. Identification of this histamine H1R/H4R-DC-CD4+ T-cell axis provides new insights into the development of peanut-induced intestinal allergic responses and for prevention and treatment of peanut allergy.


Allergens/immunology , Anaphylaxis/immunology , Arachis/adverse effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Histamine Antagonists/pharmacology , Histamine H1 Antagonists/pharmacology , Peanut Hypersensitivity/immunology , Receptors, Histamine H4/antagonists & inhibitors , Adoptive Transfer , Anaphylaxis/drug therapy , Anaphylaxis/metabolism , Anaphylaxis/pathology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cell Lineage , Chemotaxis/drug effects , Chemotaxis/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Peanut Hypersensitivity/drug therapy , Peanut Hypersensitivity/metabolism , Peanut Hypersensitivity/pathology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
...